Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
PLoS One ; 10(12): e0143182, 2015.
Article in English | MEDLINE | ID: mdl-26699128

ABSTRACT

Small interfering RNAs (siRNAs) are fundamental to the regulation of cell function. Much is known about its gene interfering mechanism, but a kinetic description of it is still lacking. Here, we derived a set of reaction-diffusion equations for multiple RNA-induced silencing complex (RISC) pathways that give quantitative temporal and spatial descriptions of the siRNA process in mammalian cell, and are able to correctly describe all salient experimentally observed patterns of sub-cellular siRNA localization, including those that, at first glance, appear irreconcilable. These results suggest siRNA sub-cellular localization mainly concerns the non-catalytic RISC-target complex, and is caused by the selectiveness of RISC-target interaction and the permeability of the nuclear membrane to siRNA strands but not to RISC-target complexes. Our method is expected to be useful in devising RNAi based cell regulation strategies.


Subject(s)
Mammals/genetics , RNA, Small Interfering/analysis , RNA-Induced Silencing Complex/physiology , Animals , Biological Transport , Kinetics , Models, Biological , RNA, Small Interfering/metabolism , RNA-Induced Silencing Complex/metabolism
2.
Mol Cell ; 56(1): 79-89, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-25280105

ABSTRACT

In animals, key functions of microRNA-induced silencing complex (miRISC) are translational repression and deadenylation followed by mRNA decay. While miRISC represses translation initiation, it is poorly understood how miRISC exerts this function. Here we assessed the effect of miRISC on synergistic recruitment of translation initiation factors to target mRNAs by using direct biochemical assays. We show that miRISC promotes eIF4AI and eIF4AII release from target mRNAs prior to dissociation of eIF4E and eIF4G in a deadenylation-independent manner. Strikingly, miRISC-induced release of eIF4AI and eIF4AII from target mRNAs and miRISC-induced inhibition of cap-dependent translation can both be counteracted by the RNA-binding protein HuD via a direct interaction of HuD with eIF4A. Furthermore, the pharmacological eIF4A inhibitor silvestrol, which locks eIF4A on mRNAs, conferred resistance to miRNA-mediated translational repression. In summary, we propose that both eIF4AI and eIF4AII are functionally important targets in miRISC-mediated translation control.


Subject(s)
Eukaryotic Initiation Factor-4A/metabolism , MicroRNAs/physiology , Models, Genetic , RNA, Messenger/metabolism , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Eukaryotic Initiation Factor-4A/genetics , HEK293 Cells , Humans , RNA-Induced Silencing Complex/physiology , Transcription Initiation, Genetic , Triterpenes/pharmacology
3.
Proc Natl Acad Sci U S A ; 111(19): 6970-5, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24778252

ABSTRACT

MicroRNAs (miRNAs) are small evolutionarily conserved regulatory RNAs that modulate mRNA stability and translation in a wide range of cell types. MiRNAs are involved in a broad array of biological processes, including cellular proliferation, differentiation, and apoptosis. To identify previously unidentified regulators of miRNA, we initiated a systematic discovery-type proteomic analysis of the miRNA pathway interactome in human cells. Six of 66 genes identified in our proteomic screen were capable of regulating lethal-7a (let-7a) miRNA reporter activity. Tripartite motif 65 (TRIM65) was identified as a repressor of miRNA activity. Detailed analysis indicates that TRIM65 interacts and colocalizes with trinucleotide repeat containing six (TNRC6) proteins in processing body-like structures. Ubiquitination assays demonstrate that TRIM65 is an ubiquitin E3 ligase for TNRC6 proteins. The combination of overexpression and knockdown studies establishes that TRIM65 relieves miRNA-driven suppression of mRNA expression through ubiquitination and subsequent degradation of TNRC6.


Subject(s)
Autoantigens/genetics , Autoantigens/metabolism , MicroRNAs/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Arabidopsis Proteins/metabolism , Glioblastoma , HEK293 Cells , HeLa Cells , Humans , Intramolecular Transferases/metabolism , Lung Neoplasms , Proteomics , RNA Stability/physiology , RNA-Induced Silencing Complex/physiology , Tripartite Motif Proteins , Ubiquitination/physiology
4.
Dig Dis Sci ; 59(4): 716-23, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24248419

ABSTRACT

Gastric cancer is one of the most common cancers and accounts for a large proportion of cancer-related deaths in the world, while the pathogenesis of it is still not clear. Epigenetic changes have been found to participate in the development and progression of gastric cancer. Epigenetic changes involve methylation of cytosines in DNA, modifications of histone, chromatin remodeling, and alterations in the expression of microRNAs. MicroRNAs, a family of small non-coding RNAs, have been demonstrated to participate in many fundamental biological processes including the carcinogenesis of gastric cancer. Previous studies have shown that the downregulation of microRNAs are often caused by the methylation in the CpG islands of microRNA promoters. Here, we have summarized the functions and molecular mechanisms of gastric cancer related methylated microRNAs in gastric carcinogenesis. We further envisage the clinical application of microRNA methylation in the early diagnosis, treatment and prognosis assessment of gastric cancer.


Subject(s)
Epigenesis, Genetic/physiology , MicroRNAs/genetics , Stomach Neoplasms/genetics , CpG Islands/genetics , CpG Islands/physiology , DNA Methylation , Down-Regulation/physiology , Humans , RNA-Induced Silencing Complex/physiology , Stomach Neoplasms/diagnosis , Stomach Neoplasms/therapy , Up-Regulation/physiology
5.
Proc Natl Acad Sci U S A ; 110(41): 16492-7, 2013 Oct 08.
Article in English | MEDLINE | ID: mdl-24067652

ABSTRACT

Piwi-interacting RNAs (piRNAs) are gonad-specific small RNAs that provide defense against transposable genetic elements called transposons. Our knowledge of piRNA biogenesis is sketchy, partly due to an incomplete inventory of the factors involved. Here, we identify Tudor domain-containing 12 (TDRD12; also known as ECAT8) as a unique piRNA biogenesis factor in mice. TDRD12 is detected in complexes containing Piwi protein MILI (PIWIL2), its associated primary piRNAs, and TDRD1, all of which are already implicated in secondary piRNA biogenesis. Male mice carrying either a nonsense point mutation (reproductive mutant 23 or repro23 mice) or a targeted deletion in the Tdrd12 locus are infertile and derepress retrotransposons. We find that TDRD12 is dispensable for primary piRNA biogenesis but essential for production of secondary piRNAs that enter Piwi protein MIWI2 (PIWIL4). Cell-culture studies with the insect ortholog of TDRD12 suggest a role for the multidomain protein in mediating complex formation with other participants during secondary piRNA biogenesis.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA Methylation/physiology , DNA Transposable Elements/physiology , Germ Cells/physiology , RNA, Small Interfering/biosynthesis , RNA-Induced Silencing Complex/physiology , Amino Acid Sequence , Animals , Blotting, Northern , Bombyx , Cloning, Molecular , Computational Biology , DNA Primers/genetics , DNA Transposable Elements/genetics , DNA, Complementary/genetics , Fluorescent Antibody Technique , Genetic Vectors/genetics , Immunoprecipitation , Male , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , RNA-Induced Silencing Complex/genetics
6.
Proteomics ; 13(3-4): 558-67, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23125164

ABSTRACT

MicroRNAs (miRNAs) are small noncoding RNAs that play important roles in posttranscriptional regulation of gene expression. Mature miRNAs associate with the RNA interference silencing complex to repress mRNA translation and/or degrade mRNA transcripts. Mass spectrometry-based proteomics has enabled identification of several core components of the canonical miRNA processing pathway and their posttranslational modifications which are pivotal in miRNA regulatory mechanisms. The use of quantitative proteomic strategies has also emerged as a key technique for experimental identification of miRNA targets by allowing direct determination of proteins whose levels are altered because of translational suppression. This review focuses on the role of proteomics and labeling strategies to understand miRNA biology.


Subject(s)
MicroRNAs/physiology , Animals , Computer Simulation , Humans , Isotope Labeling , Models, Biological , Protein Processing, Post-Translational , Proteomics , RNA Interference , RNA-Induced Silencing Complex/physiology
7.
J Cell Biol ; 198(4): 529-44, 2012 Aug 20.
Article in English | MEDLINE | ID: mdl-22891262

ABSTRACT

The microRNA (miRNA)-induced silencing complex (miRISC) controls gene expression by a posttranscriptional mechanism involving translational repression and/or promoting messenger RNA (mRNA) deadenylation and degradation. The GW182/TNRC6 (GW) family proteins are core components of the miRISC and are essential for miRNA function. We show that mammalian GW proteins have distinctive functions in the miRNA pathway, with GW220/TNGW1 being essential for the formation of GW/P bodies containing the miRISC. miRISC aggregation and formation of GW/P bodies sequestered and stabilized translationally repressed target mRNA. Depletion of GW220 led to the loss of GW/P bodies and destabilization of miRNA-targeted mRNA. These findings support a model in which the cellular localization of the miRISC regulates the fate of the target mRNA.


Subject(s)
Autoantigens/genetics , Autoantigens/metabolism , RNA Interference/physiology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , RNA-Induced Silencing Complex/physiology , Animals , Autoantigens/chemistry , Cell Line, Tumor , HEK293 Cells , HeLa Cells , Humans , MicroRNAs/metabolism , RNA Stability/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry
8.
Mol Cell ; 43(6): 892-903, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21925378

ABSTRACT

Since the establishment of a canonical animal microRNA biogenesis pathway driven by the RNase III enzymes Drosha and Dicer, an unexpected variety of alternative mechanisms that generate functional microRNAs have emerged. We review here the many Drosha-independent and Dicer-independent microRNA biogenesis strategies characterized over the past few years. Beyond reflecting the flexibility of small RNA machineries, the existence of noncanonical pathways has consequences for interpreting mutants in the core microRNA machinery. Such mutants are commonly used to assess the consequences of "total" microRNA loss, and indeed, they exhibit many overall phenotypic similarities. Nevertheless, ongoing studies reveal a growing number of settings in which alternative microRNA pathways contribute to distinct phenotypes among core microRNA biogenesis mutants.


Subject(s)
MicroRNAs/biosynthesis , Mutation , Animals , Argonaute Proteins , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/physiology , Gene Expression Regulation/physiology , Humans , MicroRNAs/chemistry , Models, Genetic , Olfactory Pathways/metabolism , RNA Splicing , RNA, Messenger/metabolism , RNA-Binding Proteins , RNA-Induced Silencing Complex/genetics , RNA-Induced Silencing Complex/physiology , Ribonuclease III/physiology
9.
Hepatology ; 53(5): 1538-48, 2011 May.
Article in English | MEDLINE | ID: mdl-21520169

ABSTRACT

UNLABELLED: There is virtually no effective treatment for advanced hepatocellular carcinoma (HCC) and novel targets need to be identified to develop effective treatment. We recently documented that the oncogene Astrocyte elevated gene-1 (AEG-1) plays a seminal role in hepatocarcinogenesis. Employing yeast two-hybrid assay and coimmunoprecipitation followed by mass spectrometry, we identified staphylococcal nuclease domain containing 1 (SND1), a nuclease in the RNA-induced silencing complex (RISC) facilitating RNAi-mediated gene silencing, as an AEG-1 interacting protein. Coimmunoprecipitation and colocalization studies confirmed that AEG-1 is also a component of RISC and both AEG-1 and SND1 are required for optimum RISC activity facilitating small interfering RNA (siRNA) and micro RNA (miRNA)-mediated silencing of luciferase reporter gene. In 109 human HCC samples SND1 was overexpressed in ≈74% cases compared to normal liver. Correspondingly, significantly higher RISC activity was observed in human HCC cells compared to immortal normal hepatocytes. Increased RISC activity, conferred by AEG-1 or SND1, resulted in increased degradation of tumor suppressor messenger RNAs (mRNAs) that are target of oncomiRs. Inhibition of enzymatic activity of SND1 significantly inhibited proliferation of human HCC cells. As a corollary, stable overexpression of SND1 augmented and siRNA-mediated inhibition of SND1 abrogated growth of human HCC cells in vitro and in vivo, thus revealing a potential role of SND1 in hepatocarcinogenesis. CONCLUSION: We unravel a novel mechanism that overexpression of AEG-1 and SND1 leading to increased RISC activity might contribute to hepatocarcinogenesis. Targeted inhibition of SND1 enzymatic activity might be developed as an effective therapy for HCC.


Subject(s)
Carcinoma, Hepatocellular/etiology , Cell Adhesion Molecules/physiology , Liver Neoplasms/etiology , Nuclear Proteins/physiology , RNA-Induced Silencing Complex/physiology , Animals , Endonucleases , Humans , Membrane Proteins , Mice , RNA-Binding Proteins , Tumor Cells, Cultured
10.
Pharmacol Ther ; 130(1): 26-37, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21256154

ABSTRACT

MicroRNAs are members of the non-protein-coding family of RNAs. They serve as regulators of gene expression by modulating the translation and/or stability of messenger RNA targets. The discovery of microRNAs has revolutionized the field of cell biology, and has permanently altered the prevailing view of a linear relationship between gene and protein expression. The increased complexity of gene regulation is both exciting and daunting, as emerging evidence supports a pervasive role for microRNAs in virtually every cellular process. This review briefly describes microRNA processing and formation of RNA-induced silencing complexes, with a focus on the role of RNA binding proteins in this process. We also discuss mechanisms for microRNA-mediated regulation of translation, particularly in dendritic spine formation and function, and the role of microRNAs in synaptic plasticity. We then discuss the evidence for altered microRNA function in cognitive brain disorders, and the effect of gene mutations revealed by single nucleotide polymorphism analysis on altered microRNA function and human disease. Further, we present evidence that altered microRNA expression in circulating fluids such as plasma/serum can correlate with, and serve as, novel diagnostic biomarkers of human disease.


Subject(s)
Brain Diseases/physiopathology , Dendritic Spines/physiology , Gene Expression Regulation , MicroRNAs/physiology , RNA, Messenger/metabolism , Synapses/physiology , Biomarkers/analysis , Brain Diseases/genetics , Cognition Disorders/physiopathology , Dendritic Spines/genetics , Humans , MicroRNAs/genetics , Neuronal Plasticity , Polymorphism, Single Nucleotide , RNA Stability , RNA, Messenger/genetics , RNA-Binding Proteins/physiology , RNA-Induced Silencing Complex/physiology
11.
Cold Spring Harb Perspect Biol ; 3(10): a003772, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-20810548

ABSTRACT

RNAi has existed at least since the divergence of prokaryotes and eukaryotes. This collection of pathways responds to a diversity of "abberant" RNAs and generally silences or eliminates genes sharing sequence content with the silencing trigger. In the canonical pathway, double-stranded RNAs are processed into small RNAs, which guide effector complexes to their targets by complementary base pairing. Many alternative routes from silencing trigger to small RNA are continuously being uncovered. Though the triggers of the pathway and the mechanisms of small RNA production are many, all RNAi-related mechanisms share Argonaute proteins as the heart of their effector complexes. These can act as self-contained silencing machines, binding directly to small RNAs, carrying out homology-based target recognition, and in some cases cleaving targets using an endogenous nuclease domain. Here, we discuss the diversity of Argonaute proteins from a structural and functional perspective.


Subject(s)
Argonaute Proteins/physiology , Evolution, Molecular , RNA, Small Interfering/physiology , Argonaute Proteins/chemistry , Argonaute Proteins/genetics , Conserved Sequence , Models, Genetic , Models, Molecular , Phylogeny , Protein Structure, Tertiary , RNA Interference , RNA, Small Interfering/chemistry , RNA-Induced Silencing Complex/chemistry , RNA-Induced Silencing Complex/physiology
12.
Life Sci ; 88(9-10): 373-83, 2011 Feb 28.
Article in English | MEDLINE | ID: mdl-21130781

ABSTRACT

Despite significant advances in treatments, cardiovascular disease (CVD) remains the leading cause of human morbidity and mortality in developed countries. The development of novel and efficient treatment strategies requires an understanding of the basic molecular mechanisms underlying cardiac function. MicroRNAs (miRNAs) are a family of small nonprotein-coding RNAs that have emerged as important regulators in cardiac and vascular developmental and pathological processes, including cardiac arrhythmia, fibrosis, hypertrophy and ischemia, heart failure and vascular atherosclerosis. The miRNA acts as an adaptor for the miRNA-induced silencing complex (miRISC) to specifically recognize and regulate particular mRNAs. Mature miRNAs recognize their target mRNAs by base-pairing interactions between nucleotides 2 and 8 of the miRNA (the seed region) and complementary nucleotides in the 3'-untranslated region (3'-UTR) of mRNAs and miRISCs subsequently inhibit gene expression by targeting mRNAs for translational repression or cleavage. In this review we summarize the basic mechanisms of action of miRNAs as they are related to cardiac arrhythmia and address the potential for miRNAs to be therapeutically manipulated in the treatment of arrhythmias.


Subject(s)
Arrhythmias, Cardiac/metabolism , MicroRNAs/physiology , Animals , Humans , Myocardium/metabolism , RNA Interference/physiology , RNA, Messenger/metabolism , RNA-Induced Silencing Complex/physiology
13.
Nat Genet ; 43(2): 153-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21186352

ABSTRACT

Canalization, also known as developmental robustness, describes an organism's ability to produce the same phenotype despite genotypic variations and environmental influences. In Drosophila, Hsp90, the trithorax-group proteins and transposon silencing have been previously implicated in canalization. Despite this, the molecular mechanism underlying canalization remains elusive. Here using a Drosophila eye-outgrowth assay sensitized by the dominant Kr(irregular facets-1)(Kr(If-1)) allele, we show that the Piwi-interacting RNA (piRNA) pathway, but not the short interfering RNA or micro RNA pathway, is involved in canalization. Furthermore, we isolated a protein complex composed of Hsp90, Piwi and Hop, the Hsp70/Hsp90 organizing protein homolog, and we demonstrated the function of this complex in canalization. Our data indicate that Hsp90 and Hop regulate the piRNA pathway through Piwi to mediate canalization. Moreover, they point to epigenetic silencing of the expression of existing genetic variants and the suppression of transposon-induced new genetic variation as two major mechanisms underlying piRNA pathway-mediated canalization.


Subject(s)
Drosophila Proteins/physiology , HSP90 Heat-Shock Proteins/metabolism , RNA-Induced Silencing Complex/physiology , Alleles , Animals , Argonaute Proteins , DNA Transposable Elements , Drosophila Proteins/genetics , Drosophila melanogaster , Electrophoresis, Gel, Two-Dimensional , Epigenesis, Genetic , Female , Gene Silencing , Genetic Variation , Green Fluorescent Proteins/metabolism , Male , Ovary/metabolism , Phenotype , RNA-Induced Silencing Complex/genetics
14.
Crit Rev Eukaryot Gene Expr ; 20(2): 141-8, 2010.
Article in English | MEDLINE | ID: mdl-21133843

ABSTRACT

Processing of the pre-microRNA (pre-miRNA) through Dicer1 generates a miRNA duplex, consisting of a miRNA and miRNA* strand (also termed guide strand and passenger strand, respectively). Despite the general consensus that miRNA*s have no regulatory activity, recent publications have provided evidence that the abundance, possible function, and physiological relevance of miRNA*s have been underestimated. This review provides an account of our current understanding of miRNA* origination and activity, mounting evidence for their unique functions and regulatory mechanisms, and examples of specific miRNA*s from the literature.


Subject(s)
MicroRNAs/physiology , RNA-Induced Silencing Complex/physiology , Animals , Humans
15.
Nat Struct Mol Biol ; 17(10): 1169-74, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20924405

ABSTRACT

MicroRNAs (miRNAs) suppress gene expression by inhibiting translation, promoting mRNA decay or both. Each miRNA may regulate hundreds of genes to control the cell's response to developmental and other environmental cues. The best way to understand the function of a miRNA is to identify the genes that it regulates. Target gene identification is challenging because miRNAs bind to their target mRNAs by partial complementarity over a short sequence, suppression of an individual target gene is often small, and the rules of targeting are not completely understood. Here we review computational and experimental approaches to the identification of miRNA-regulated genes. The examination of changes in gene expression that occur when miRNA expression is altered and biochemical isolation of miRNA-associated transcripts complement target prediction algorithms. Bioinformatic analysis of over-represented pathways and nodes in protein-DNA interactomes formed from experimental candidate miRNA gene target lists can focus attention on biologically significant target genes.


Subject(s)
Computational Biology/methods , Gene Expression Regulation , MicroRNAs/physiology , RNA-Induced Silencing Complex/physiology , 3' Untranslated Regions , Algorithms , Animals , Autoantigens/physiology , Base Pairing , Base Sequence , Binding Sites , Caenorhabditis elegans/genetics , Databases, Genetic , Gene Knockdown Techniques , Gene Regulatory Networks , Humans , Mice , Oligonucleotide Array Sequence Analysis , Proteomics , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/physiology
16.
Neurosci Lett ; 471(3): 185-8, 2010 Mar 08.
Article in English | MEDLINE | ID: mdl-20109527

ABSTRACT

RNA-Induced Silencing Complex (RISC) mediates post-transcriptional control of gene expression and contains Argonaute 2 (AGO2) protein as a central effector of cleavage or inhibition of mRNA translation. In the brain, the RISC pathway is involved in neuronal functions, such as synaptic development and local protein synthesis, which are potentially critical for memory. In this study, we examined the role of RISC in memory formation in rodents, by silencing AGO2 expression in dorsal hippocampus of C57BL/6 mice and submitting animals to hippocampus-related tasks. One week after surgery, AGO2 downregulation impaired both short-term and long-term contextual fear memories. Conversely, no long-lasting effects were observed three weeks after surgery, when AGO2 levels were re-established. These results show that altered RISC activity severely affects learning and memory processes in rodents.


Subject(s)
Eukaryotic Initiation Factor-2/biosynthesis , Hippocampus/metabolism , Memory , RNA-Induced Silencing Complex/physiology , Animals , Argonaute Proteins , Conditioning, Psychological , Down-Regulation , Eukaryotic Initiation Factor-2/genetics , Fear , Gene Silencing , Memory, Short-Term , Mice , Mice, Inbred C57BL , Time Factors
17.
PLoS One ; 4(10): e7618, 2009 Oct 27.
Article in English | MEDLINE | ID: mdl-19888420

ABSTRACT

Fragile X Syndrome is caused by the silencing of the Fragile X Mental Retardation gene (FMR1). Regulating dosage of FMR1 levels is critical for proper development and function of the nervous system and germ line, but the pathways responsible for maintaining normal expression levels are less clearly defined. Loss of Drosophila Fragile X protein (dFMR1) causes several behavioral and developmental defects in the fly, many of which are analogous to those seen in Fragile X patients. Over-expression of dFMR1 also causes specific neuronal and behavioral abnormalities. We have found that Argonaute2 (Ago2), the core component of the small interfering RNA (siRNA) pathway, regulates dfmr1 expression. Previously, the relationship between dFMR1 and Ago2 was defined by their physical interaction and co-regulation of downstream targets. We have found that Ago2 and dFMR1 are also connected through a regulatory relationship. Ago2 mediated repression of dFMR1 prevents axon growth and branching defects of the Drosophila neuromuscular junction (NMJ). Consequently, the neurogenesis defects in larvae mutant for both dfmr1 and Ago2 mirror those in dfmr1 null mutants. The Ago2 null phenotype at the NMJ is rescued in animals carrying an Ago2 genomic rescue construct. However, animals carrying a mutant Ago2 allele that produces Ago2 with significantly reduced endoribonuclease catalytic activity are normal with respect to the NMJ phenotypes examined. dFMR1 regulation by Ago2 is also observed in the germ line causing a multiple oocyte in a single egg chamber mutant phenotype. We have identified Ago2 as a regulator of dfmr1 expression and have clarified an important developmental role for Ago2 in the nervous system and germ line that requires dfmr1 function.


Subject(s)
Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Drosophila melanogaster/genetics , Fragile X Mental Retardation Protein/metabolism , Oogenesis/physiology , RNA-Induced Silencing Complex/physiology , Animals , Argonaute Proteins , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Female , Fragile X Mental Retardation Protein/genetics , Gene Expression Regulation , Gene Silencing , Microscopy, Fluorescence/methods , Models, Biological , Motor Neurons/metabolism , Nervous System/metabolism , Neurons/metabolism , Phenotype , RNA, Small Interfering/metabolism
18.
PLoS One ; 4(7): e6182, 2009 Jul 08.
Article in English | MEDLINE | ID: mdl-19584931

ABSTRACT

Heterochromatin formation plays an important role in gene regulation and the maintenance of genome integrity. Here we present results from a study of the D. melanogaster gene vig, encoding an RNAi complex component and its homolog vig2 (CG11844) that support their involvement in heterochromatin formation and/or maintenance. Protein null mutations vig(EP812) and vig2(PL470) act as modifiers of Position Effect Variegation (PEV). VIG and Vig2 are present in polytene chromosomes and partially overlap with HP1. Quantitative immunoblots show depletion of HP1 and HP2 (large isoform) in isolated nuclei from the vig(EP812) mutant. The vig2(PL470) mutant strain demonstrates a decreased level of H3K9me2. Pull-down experiments using antibodies specific to HP1 recovered both VIG and Vig2. The association between HP1 and both VIG and Vig2 proteins depends on an RNA component. The above data and the developmental profiles of the two genes suggest that Vig2 may be involved in heterochromatin targeting and establishment early in development, while VIG may have a role in stabilizing HP1/HP2 chromatin binding during later stages.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/physiology , Heterochromatin/metabolism , RNA-Induced Silencing Complex/physiology , Animals , Base Sequence , Blotting, Western , DNA Primers , Gene Expression Profiling , Gene Expression Regulation, Developmental , Immunoprecipitation , Reverse Transcriptase Polymerase Chain Reaction
19.
Mol Cell ; 34(2): 155-67, 2009 Apr 24.
Article in English | MEDLINE | ID: mdl-19394293

ABSTRACT

RNA interference (RNAi) plays a pivotal role in the formation of heterochromatin at the fission yeast centromeres. The RNA-induced transcriptional silencing (RITS) complex, composed of heterochromatic small interfering RNAs (siRNAs), the siRNA-binding protein Ago1, the chromodomain protein Chp1, and the Ago1/Chp1-interacting protein Tas3, provides a physical tether between the RNAi and heterochromatin assembly pathways. Here, we report the structural and functional characterization of a C-terminal Tas3 alpha-helical motif (TAM), which self-associates into a helical polymer and is required for cis spreading of RITS in centromeric DNA regions. Site-directed mutations of key residues within the hydrophobic monomer-monomer interface disrupt Tas3-TAM polymeric self-association in vitro and result in loss of gene silencing, spreading of RITS, and a dramatic reduction in centromeric siRNAs in vivo. These results demonstrate that, in addition to the chromodomain of Chp1 and siRNA-loaded Ago1, Tas3 self-association is required for RITS spreading and efficient heterochromatic gene silencing at centromeric repeat regions.


Subject(s)
Carrier Proteins/physiology , Gene Silencing/physiology , Heterochromatin/genetics , RNA-Induced Silencing Complex/metabolism , Schizosaccharomyces pombe Proteins/physiology , Schizosaccharomyces/genetics , Amino Acid Motifs , Carrier Proteins/chemistry , Carrier Proteins/genetics , Centromere/genetics , Centromere/metabolism , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation , Chromatography, Gel , Crystallography, X-Ray , Histones/metabolism , Lysine/metabolism , Methylation , Models, Molecular , Mutagenesis, Site-Directed , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , RNA-Induced Silencing Complex/physiology , Schizosaccharomyces/enzymology , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces pombe Proteins/genetics
20.
Mol Cell ; 34(1): 58-67, 2009 Apr 10.
Article in English | MEDLINE | ID: mdl-19268617

ABSTRACT

microRNAs induce translational repression by binding to partially complementary sites on their target mRNAs. We have established an in vitro system that recapitulates translational repression mediated by the two Drosophila Argonaute (Ago) subfamily proteins, Ago1 and Ago2. We find that Ago1-RISC (RNA-induced silencing complex) represses translation primarily by ATP-dependent shortening of the poly(A) tail of its mRNA targets. Ago1-RISC can also secondarily block a step after cap recognition. In contrast, Ago2-RISC competitively blocks the interaction of eIF4E with eIF4G and inhibits the cap function. Our finding that the two Ago proteins in flies regulate translation by different mechanisms may reconcile previous, contradictory explanations for how miRNAs repress protein synthesis.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/genetics , Protein Biosynthesis/physiology , RNA-Induced Silencing Complex/physiology , Adenosine Triphosphate/physiology , Animals , Argonaute Proteins , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Eukaryotic Initiation Factor-4E/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Eukaryotic Initiation Factors , RNA/metabolism , RNA-Induced Silencing Complex/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...